Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 202
Filtrar
1.
J Neurosci Res ; 99(9): 2305-2317, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34115908

RESUMO

The consolidation of new memories into long-lasting memories is multistage process characterized by distinct temporal dynamics. However, our understanding on the initial stage of transformation of labile memory of recent experience into stable memory remains elusive. Here, with the use of rats and mice overexpressing a memory enhancer called regulator of G protein signaling 14 of 414 amino acids (RGS14414 ) as a tool, we show that the expression of RGS14414 in male rats' perirhinal cortex (PRh), which is a brain area crucial for object recognition memory (ORM), enhanced the ORM to the extent that it caused the conversion of labile short-term ORM (ST-ORM) expected to last for 40 min into stable long-term ORM (LT-ORM) traceable after a delay of 24 hr, and that the temporal window of 40 to 60 min after object exposure not only was key for this conversion but also was the time frame when a surge in 14-3-3ζ protein was observed. A knockdown of 14-3-3ζ gene abrogated both the increase in 14-3-3ζ protein and the formation of LT-ORM. Furthermore, this 14-3-3ζ upregulation increased brain-derived growth factor (BDNF) levels in the time frame of 60 min and 24 hr and 14-3-3ζ knockdown decreased the BDNF levels, and a deletion of BDNF gene produced loss in mice ability to form LT-ORM. Thus, within 60 min of object exposure, 14-3-3ζ facilitated the conversion of labile ORM into stable ORM, whereas beyond the 60 min, it mediated the consolidation of the stable memory into long-lasting ORM by regulating BDNF signaling.


Assuntos
Proteínas 14-3-3/biossíntese , Memória de Longo Prazo/fisiologia , Memória de Curto Prazo/fisiologia , Reconhecimento Psicológico/fisiologia , Proteínas 14-3-3/genética , Animais , Fator Neurotrófico Derivado do Encéfalo/deficiência , Fator Neurotrófico Derivado do Encéfalo/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Ratos , Ratos Wistar , Percepção Visual/fisiologia
2.
Brain Res ; 1754: 147254, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33422542

RESUMO

Schizophrenia is a neurodevelopmental psychiatric disorder, encompassing genetic and environmental risk factors. For several decades, investigators have been implementing the use of lesions of the neonatal rodent hippocampus to model schizophrenia, resulting in a broad spectrum of adult schizophrenia-related behavioral changes. Despite the extensive use of these proposed animal models of schizophrenia, the mechanisms by which these lesions result in schizophrenia-like behavioral alterations remain unclear. Here we provide in vivo evidence that transient pharmacological inactivation of the hippocampus via tetrodotoxin microinjections or a genetic reduction in brain derived neurotrophic factor (BDNF) protein levels (BDNF+/- rats) lead to global DNA hypomethylation, disrupted maturation of the neuronal nucleus and aberrant acoustic startle response in the adult rat. The similarity between the effects of the two treatments strongly indicate that BDNF signaling is involved in effects obtained after the TTX microinjections. These findings may shed light on the cellular mechanisms underlying the phenotypical features of neonatal transient inhibition of the hippocampus as a preclinical model of schizophrenia and suggest that BDNF signaling represents a target pathway for development of novel treatment therapies.


Assuntos
Comportamento Animal/fisiologia , Fator Neurotrófico Derivado do Encéfalo/deficiência , Metilação de DNA/fisiologia , DNA/metabolismo , Hipocampo/metabolismo , Animais , Animais Recém-Nascidos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Neurônios/metabolismo , Ratos , Reflexo de Sobressalto/genética , Reflexo de Sobressalto/fisiologia , Esquizofrenia/genética , Esquizofrenia/metabolismo
3.
Cell Mol Neurobiol ; 41(4): 751-763, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-32445041

RESUMO

Exposure to noise produces cognitive and emotional disorders, and recent studies have shown that auditory stimulation or deprivation affects hippocampal function. Previously, we showed that exposure to high-intensity sound (110 dB, 1 min) strongly inhibits Schaffer-CA1 long-term potentiation (LTP). Here we investigated possible mechanisms involved in this effect. We found that exposure to 110 dB sound activates c-fos expression in hippocampal CA1 and CA3 neurons. Although sound stimulation did not affect glutamatergic or GABAergic neurotransmission in CA1, it did depress the level of brain-derived neurotrophic factor (BDNF), which is involved in promoting hippocampal synaptic plasticity. Moreover, perfusion of slices with BDNF rescued LTP in animals exposed to sound stimulation, whereas BDNF did not affect LTP in sham-stimulated rats. Furthermore, LM22A4, a TrkB receptor agonist, also rescued LTP from sound-stimulated animals. Our results indicate that depression of hippocampal BDNF mediates the inhibition of LTP produced by high-intensity sound stimulation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/deficiência , Hipocampo/fisiologia , Potenciação de Longa Duração , Som , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Região CA1 Hipocampal/fisiologia , Ácido Glutâmico/metabolismo , Potenciação de Longa Duração/fisiologia , Masculino , Proteínas Proto-Oncogênicas c-fos/metabolismo , Células Piramidais/metabolismo , Ratos Wistar , Sinapses/fisiologia , Transmissão Sináptica , Ácido gama-Aminobutírico/metabolismo
4.
JCI Insight ; 6(3)2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33351789

RESUMO

Paucity of the glucose transporter-1 (Glut1) protein resulting from haploinsufficiency of the SLC2A1 gene arrests cerebral angiogenesis and disrupts brain function to cause Glut1 deficiency syndrome (Glut1 DS). Restoring Glut1 to Glut1 DS model mice prevents disease, but the precise cellular sites of action of the transporter, its temporal requirements, and the mechanisms linking scarcity of the protein to brain cell dysfunction remain poorly understood. Here, we show that Glut1 functions in a cell-autonomous manner in the cerebral microvasculature to affect endothelial tip cells and, thus, brain angiogenesis. Moreover, brain endothelial cell-specific Glut1 depletion not only triggers a severe neuroinflammatory response in the Glut1 DS brain, but also reduces levels of brain-derived neurotrophic factor (BDNF) and causes overt disease. Reduced BDNF correlated with fewer neurons in the Glut1 DS brain. Controlled depletion of the protein demonstrated that brain pathology and disease severity was greatest when Glut1 scarcity was induced neonatally, during brain angiogenesis. Reducing Glut1 at later stages had mild or little effect. Our results suggest that targeting brain endothelial cells during early development is important to ensure proper brain angiogenesis, prevent neuroinflammation, maintain BDNF levels, and preserve neuron numbers. This requirement will be essential for any disease-modifying therapeutic strategy for Glut1 DS.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/metabolismo , Transportador de Glucose Tipo 1/deficiência , Transportador de Glucose Tipo 1/metabolismo , Proteínas de Transporte de Monossacarídeos/deficiência , Animais , Animais Recém-Nascidos , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Encéfalo/patologia , Fator Neurotrófico Derivado do Encéfalo/deficiência , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Erros Inatos do Metabolismo dos Carboidratos/genética , Erros Inatos do Metabolismo dos Carboidratos/patologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Técnicas de Silenciamento de Genes , Transportador de Glucose Tipo 1/genética , Haploinsuficiência , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Neovascularização Fisiológica/genética , Neurônios/metabolismo , Neurônios/patologia , Fenótipo
5.
Exp Neurol ; 334: 113438, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32822705

RESUMO

Peripheral nerve injuries can significantly reduce quality of life. While some recover, most do not recover fully, resulting in neuropathic pain and loss of sensation and motor function. Research on the mechanisms of peripheral nerve regeneration could elucidate poor patient outcomes and potential treatments. This study was designed to determine if brain derived neurotrophic factor (BDNF) is necessary for pudendal nerve regeneration and functional recovery. Peripheral administration of tyrosine kinase B functional chimera (TrkB) was used to inhibit the BDNF regenerative pathway. Female Sprague-Dawley rats received tyrosine kinase B functional chimera (TrkB) or saline after a pudendal nerve crush (PNC) or Sham PNC and were divided into three groups: Sham PNC, PNC + Saline, and PNC + TrkB. Seven days after injury, relative ßII tubulin expression (1.0 ± 0.2) was significantly decreased after PNC + TrkB compared to PNC + saline (2.9 ± 1.0). Three weeks after injury, BDNF plasma concentration (1320.8 ± 278.1 pg/ml) was significantly reduced in PNC + TrkB compared to PNC + saline rats (2053.4 ± 211.0 pg/ml). Pudendal nerve motor branch firing rate (54.0 ± 9.5 Hz) was significantly decreased in the PNC + TrkB group compared to the PNC + saline group (120.4 ± 17.1 Hz); while nerve firing rate of the PNC + saline group was not significantly different from sham PNC rats (121.8 ± 26.6 Hz). This study demonstrated that peripheral administration of TrkB bound free BDNF and inhibited the regenerative response after PNC. BDNF is necessary for normal PN motor branch recovery after PNC.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/antagonistas & inibidores , Fator Neurotrófico Derivado do Encéfalo/deficiência , Regeneração Nervosa/fisiologia , Nervo Pudendo/lesões , Nervo Pudendo/fisiologia , Animais , Feminino , Compressão Nervosa/efeitos adversos , Compressão Nervosa/métodos , Regeneração Nervosa/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor trkB/farmacologia
6.
Front Immunol ; 11: 1357, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32676082

RESUMO

Streptococcus pneumoniae meningitis is a life-threatening bacterial infection of the central nervous system (CNS), and its unfavorable prognosis usually results from an intense inflammatory response. Recent studies have shown that brain-derived neurotrophic factor (BDNF) mediates anti-inflammatory and neuroprotective effects in CNS diseases; however, the distinct contribution of BDNF to pneumococcal meningitis (PM) remains unknown. In this study, we sought to investigate the effects of endogenous BDNF on the inflammatory response and brain damage in experimental PM. We used Camk2a-CreERT2 mice to delete Bdnf from the cerebral cortex and hippocampus, and meningitis was induced by intracisternal infection with S. pneumoniae. Clinical parameters were assessed during acute meningitis. At 24 h post-infection, histopathology, neutrophil granulocytes infiltration, and microglia/macrophage proliferation of brain tissues were evaluated. Additionally, cortical damage and hippocampal apoptosis were assessed using Nissl staining and terminal deoxynucleotidyl transferase dUTP-nick-end labeling (TUNEL), respectively. Pro-inflammatory cytokine levels were determined using real-time polymerase chain reaction (RT-PCR). Key molecules associated with the related signaling pathways were analyzed by RT-PCR and western blot. To investigate the role of microglia/macrophage in infected BDNF conditional knockout mice, GW2580 was used for microglia/macrophage depletion. Here, we, for the first time, found that BDNF conditional knockouts exhibited more profound clinical impairment, pathological severity, and neuron injury and enhanced microglia/macrophage proliferation than were observed in their littermate controls. Furthermore, the BDNF conditional knockouts showed an obviously increase in the expression of pro-inflammatory factors (Tnf-α, Il-1ß, and Il-6). Mechanistically, loss of BDNF activated TLR2- and NOD2-mediated downstream nuclear factor kappa B (NF-κB) p65 and p38 mitogen-activated protein kinase (MAPK) pathways associated with S. pneumoniae infection. Furthermore, targeted depletion of microglia/macrophage population decreased the resistance of mice to PM with diminishing neuroinflammation in BDNF conditional knockouts. Our findings suggest that loss of BDNF may enhance the inflammatory response and contribute to brain injury during PM at least partially by modulating TLR2- and NOD2-mediated signaling pathways, thereby providing a potential therapeutic target for future interventions in bacterial meningitis pathologies.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/imunologia , Inflamação/imunologia , Inflamação/patologia , Meningite Pneumocócica/imunologia , Meningite Pneumocócica/patologia , Animais , Lesões Encefálicas/imunologia , Lesões Encefálicas/patologia , Fator Neurotrófico Derivado do Encéfalo/deficiência , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/imunologia
7.
J Neurochem ; 154(1): 41-55, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32222968

RESUMO

Deficiency of activity-induced expression of brain-derived neurotrophic factor (BDNF) disturbs neurotransmitter gene expression. Enriched environment treatment (EET) ameliorates the defects. However, how BDNF deficiency and EET affect the neurotransmitter gene expression differently across ages remains unclear. We addressed this question by determining the neurotransmitter gene expression across three life stages in wild-type and activity-dependent BDNF-deficient (KIV) mice. Mice received 2-months of standard control treatment (SCT) or EET at early-life development (ED: 0-2 months), young adulthood (2-4 months), and old adulthood (12-14 months) (N = 16/group). Half of these mice received additional 1-month SCT to examine persisting EET effects. High-throughput quantitative reverse transcription polymerase chain reaction measured expression of 81 genes for dopamine, adrenaline, serotonin, gamma aminobutyric acid, glutamate, acetylcholine, and BDNF systems in the frontal cortex (FC) and hippocampus. Results revealed that BDNF deficiency mostly reduced neurotransmitter gene expression, greatest at ED in the FC. EET increased expression of a larger number of genes at ED than adulthood, particularly in the KIV FC. Many genes down-regulated in KIV mice were up-regulated by EET, which persisted when EET was provided at ED (e.g., 5-hydroxytryptamine (serotonin) transporter [5HTT], ADRA1D, GRIA3, GABRA5, GABBR2). In both the regions, BDNF deficiency decreased the density of gene co-expression network specifically at ED, while EET increased the density and hub genes (e.g., GAT1, GABRG3, GRIN1, CHRNA7). These results suggest that BDNF deficiency, which occurs under chronic stress, causes neurotransmitter dysregulations prominently at ED, particularly in the FC. EET at ED may be most effective to normalize the dysregulations, providing persisting effects later in life. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. More information about the Open Science badges can be found at https://cos.io/our-services/open-science-badges/.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/deficiência , Encéfalo/metabolismo , Expressão Gênica/fisiologia , Neurogênese/fisiologia , Neurotransmissores/metabolismo , Animais , Animais Recém-Nascidos , Abrigo para Animais , Camundongos , Camundongos Endogâmicos C57BL
8.
Mol Metab ; 31: 109-123, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31918913

RESUMO

OBJECTIVE: Brown adipose tissue (BAT)-mediated thermogenesis plays a key role in energy homeostasis and the maintenance of body temperature. Previous work suggests that brain-derived neurotrophic factor (BDNF) is involved in BAT thermogenesis, but the underlying neural circuits and molecular mechanism remain largely unknown. This is in part due to the difficulties in manipulating BDNF expression in different brain regions through different promoters and the lack of tools to identify neurons in the brain specifically involved in BAT thermogenesis. METHODS: We have created several lines of mutant mice in which BDNF transcription from a specific promoter was selectively disrupted by replacing Bdnf with green fluorescent protein (GFP; Bdnf-e1, -e4, and -e6-/- mice). As such, cells expressing Bdnf-e1, -e4, or -e6 were labeled with GFP. To identify BAT-connected thermogenesis neurons in brain, we applied the retrograde pseudorabies virus labeling method from BAT. We also used chemogenetic tools to manipulate specific neurons coupled with BAT temperature recording. Moreover, we developed a new TrkB agonist antibody to rescue the BAT thermogenesis deficits. RESULTS: We show that selective disruption of Bdnf expression from promoter 1 (Bdnf-e1) resulted in severe obesity and deficits of BAT-mediated thermogenesis. Body temperature response to cold was impaired in Bdnf-e1-/- mice. BAT expression of Ucp1 and Pcg1a, genes known to regulate thermogenesis, was also reduced, accompanying a decrease in the sympathetic activity of BAT. Staining of cells expressing Bdnf-e1 transcript, combined with transsynaptic, retrograde-tracing labeling of BAT-connected neurons, identified a group of excitatory neurons in lateral hypothalamus (LH) critical for thermogenesis regulation. Moreover, an adaptive thermogenesis defect in Bdnf-e1-/- mice was rescued by injecting an agonistic antibody for TrkB, the BDNF receptor, into LH. Remarkably, activation of the excitatory neurons (VGLUT2+) in LH through chemogenetic tools resulted in a rise of BAT temperature. CONCLUSIONS: These results reveal a specific role of BDNF promoter I in thermogenesis regulation and define a small subset of neurons in LH that contribute to such regulation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Ácido Glutâmico/metabolismo , Região Hipotalâmica Lateral/metabolismo , Neurônios/metabolismo , Termogênese , Animais , Fator Neurotrófico Derivado do Encéfalo/deficiência , Fator Neurotrófico Derivado do Encéfalo/genética , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes
9.
Behav Brain Res ; 377: 112223, 2020 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-31518662

RESUMO

Brain-derived neurotrophic factor (BDNF) has been implicated in cognition and the effects of chronic stress. We have previously shown in mice that chronic adolescent treatment with corticosterone (CORT), to simulate stress, resulted in spatial memory deficits and markedly elevated levels of the N-methyl-D-aspartate (NMDA) receptor subunit NR2B in adult male BDNF heterozygous mice (BDNF+/-), but not in wildtype controls (WT) or females. The aim of the present study was to further characterize this 'two hit' model, including whether these effects are long-lasting. CORT treatment was delivered in the drinking water from 6 to 9 weeks of age. As previously demonstrated, male BDNF+/- mice treated with CORT presented with a deficit in spatial memory at 11 weeks of age. However, this deficit was not maintained at 15 weeks of age. Conversely, male WT treated with CORT developed a deficit only at 15 weeks of age. There were no significant gene-environment interactions in female mice at any time point. CORT treatment caused a modest, but significant increase in NR2B levels which was independent of genotype. These results show marked age-dependent and sex-dependent effects of CORT on behaviour which are different in BDNF+/- mice than in controls.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Corticosterona/farmacologia , Glucocorticoides/farmacologia , Transtornos da Memória , Memória de Longo Prazo , Receptores de N-Metil-D-Aspartato/metabolismo , Memória Espacial , Estresse Psicológico , Fatores Etários , Animais , Fator Neurotrófico Derivado do Encéfalo/deficiência , Corticosterona/administração & dosagem , Modelos Animais de Doenças , Feminino , Interação Gene-Ambiente , Glucocorticoides/administração & dosagem , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/metabolismo , Transtornos da Memória/fisiopatologia , Memória de Longo Prazo/efeitos dos fármacos , Memória de Longo Prazo/fisiologia , Camundongos , Camundongos Knockout , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Caracteres Sexuais , Memória Espacial/efeitos dos fármacos , Memória Espacial/fisiologia , Estresse Psicológico/induzido quimicamente , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia
10.
Cell Rep ; 29(6): 1579-1593.e6, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31693897

RESUMO

Pain involves an intrinsically dynamic connectome characterized by fluctuating spontaneous brain activity and continuous neuroplastic changes of relevant circuits. Activity in the hippocampus-medial prefrontal cortex (mPFC) pathway has been suggested to correlate with spontaneous pain and pain chronicity, but causal evidence is lacking. Here we combine longitudinal in vivo electrophysiological recording with behavioral testing and show that persistent spontaneous pain disrupts ventral hippocampal CA1-infralimbic cortex (vCA1-IL) connectivity and hippocampal modulation of IL neuronal activity in rats with peripheral inflammation. Chemo- and optogenetic rescue of vCA1-IL dysfunction relieves spontaneous pain. Circuit-specific overexpression of brain-derived neurotrophic factor (BDNF) in vCA1-IL reverses electrophysiological changes, relieves spontaneous pain, and accelerates overall recovery from inflammatory pain. Our work identifies a neural pathway that specifically correlates with spontaneous pain and supports the significance of using a circuit dynamics-based strategy for more comprehensive understanding of circuitry mechanisms underlying chronic pain.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Região CA1 Hipocampal/fisiologia , Dor Crônica/fisiopatologia , Hipocampo/metabolismo , Córtex Pré-Frontal/fisiologia , Células Piramidais/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/deficiência , Fator Neurotrófico Derivado do Encéfalo/genética , Dor Crônica/induzido quimicamente , Dor Crônica/genética , Dor Crônica/metabolismo , Adjuvante de Freund/farmacologia , Inflamação/metabolismo , Masculino , Vias Neurais/fisiologia , Plasticidade Neuronal , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/metabolismo , Células Piramidais/metabolismo , Ratos
11.
Eur Arch Psychiatry Clin Neurosci ; 269(4): 439-447, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29453493

RESUMO

The brain-derived neurotrophic factor (BDNF) is a major proliferative agent in the nervous system. Both BDNF-deficiency and perinatal hypoxia represent genetic/environmental risk factors for schizophrenia. Moreover, a decreased BDNF response to birth hypoxia was associated with the disease. BDNF expression is influenced by neuronal activity and environmental conditions such as hypoxia. Thus, it may partake in neuroprotective and reparative mechanisms in acute or chronic neuronal insults. However, the interaction of hypoxia and BDNF is insufficiently understood and the behavioral outcome unknown. Therefore, we conducted a battery of behavioral tests in a classical model of chronic early postnatal mild hypoxia (10% O2), known to significantly impair brain development, in BDNF-deficient mice. We found selective deficits in measures associated with sensorimotor gating, namely enhanced acoustic startle response (ASR) and reduced prepulse inhibition (PPI) of ASR in BDNF-deficient mice. Unexpectedly, the alterations of sensorimotor gating were caused only by BDNF-deficiency alone, whereas hypoxia failed to evoke severe deficits and even leads to a milder phenotype in BDNF-deficient mice. As deficits in sensorimotor gating are present in schizophrenia and animal models of the disease, our results are of relevance regarding the involvement of BDNF in its pathogenesis. On the other hand, they suggest that the effect of perinatal hypoxia on long-term brain abnormalities is complex, ranging from protective to deleterious actions, and may critically depend on the degree of hypoxia. Therefore, future studies may refine existing hypoxia protocols to better understand neurodevelopmental consequences associated with schizophrenia.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/deficiência , Hipóxia/fisiopatologia , Inibição Pré-Pulso/fisiologia , Esquizofrenia/fisiopatologia , Filtro Sensorial/fisiologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL
12.
Biochem Biophys Res Commun ; 503(4): 2690-2697, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30100066

RESUMO

Optineurin (OPTN) mutations are linked to glaucoma pathology and E50K mutation shows massive cell death in photoreceptor cells and retinal ganglion cells. However, little is known about E50K-mediated mitochondrial dysfunction in photoreceptor cell degeneration. We here show that overexpression of E50K expression triggered BDNF deficiency, leading to Bax activation in RGC-5 cells. BDNF deficiency induced mitochondrial dysfunction by decreasing mitochondrial maximal respiration and reducing intracellular ATP level in RGC-5 cells. However, BDNF deficiency did not alter mitochondrial dynamics. Also, BDNF deficiency resulted in LC3-mediated mitophagosome formation in RGC-5 cells. These results strongly suggest that E50K-mediated BDNF deficiency plays a critical role in compromised mitochondrial function in glaucomatous photoreceptor cell degeneration.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Proteínas Associadas aos Microtúbulos/genética , Mitocôndrias/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Fator de Transcrição TFIIIA/genética , Proteína X Associada a bcl-2/genética , Trifosfato de Adenosina/biossíntese , Substituição de Aminoácidos , Animais , Fator Neurotrófico Derivado do Encéfalo/deficiência , Linhagem Celular , Regulação da Expressão Gênica , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/patologia , Mutação , Fosforilação Oxidativa , Fagossomos/metabolismo , Fagossomos/patologia , Células Fotorreceptoras de Vertebrados/patologia , Ratos , Transdução de Sinais , Fator de Transcrição TFIIIA/metabolismo , Proteína X Associada a bcl-2/metabolismo
13.
Cell Rep ; 22(13): 3401-3408, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29590610

RESUMO

Transcriptional analysis of brain tissue from people with molecularly defined causes of obesity may highlight disease mechanisms and therapeutic targets. We performed RNA sequencing of hypothalamus from individuals with Prader-Willi syndrome (PWS), a genetic obesity syndrome characterized by severe hyperphagia. We found that upregulated genes overlap with the transcriptome of mouse Agrp neurons that signal hunger, while downregulated genes overlap with the expression profile of Pomc neurons activated by feeding. Downregulated genes are expressed mainly in neuronal cells and contribute to neurogenesis, neurotransmitter release, and synaptic plasticity, while upregulated, predominantly microglial genes are involved in inflammatory responses. This transcriptional signature may be mediated by reduced brain-derived neurotrophic factor expression. Additionally, we implicate disruption of alternative splicing as a potential molecular mechanism underlying neuronal dysfunction in PWS. Transcriptomic analysis of the human hypothalamus may identify neural mechanisms involved in energy homeostasis and potential therapeutic targets for weight loss.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/deficiência , Jejum/fisiologia , Hipotálamo/metabolismo , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Humanos , Camundongos , Obesidade/metabolismo , Síndrome de Prader-Willi/patologia , Transcriptoma
14.
Rev. Hosp. Ital. B. Aires (2004) ; 38(1): 5-10, mar. 2018. tab.
Artigo em Inglês | LILACS | ID: biblio-1023455

RESUMO

The dysfunction in the serotoninergic neurotransmission has been classically associated with major depressive disorder (MDD); however, other pathways and processes seem to have a role in this illness, such as neurogenesis and related molecules: the Brain-Derived Neurotrophic Factor (BDNF) and the Apolipoprotein E (APOE). There are many reports that indicate an association between certain polymorphism in these genes and MDD. The aim of our study was to analyze the possible association between MDD and polymorphisms in HTR2A (5-hydroxytryptamine receptor 2A), BDNF and APOE genes in a sample of the Argentinean population previously studied for 2 polymorphisms in SLC6A4 (Solute Carrier Family 6 Member 4) gene. Five polymorphisms were studied (rs6311 and rs6313 in HTR2A; rs429358 and rs7412 in APOE, and rs6265 in BDNF) in 95 MDD patients and 107 non-related controls. No statistically significant differences were observed between groups when analyzing the association with a single marker using logistic regression; however, when a possible combinatory effect of the polymorphisms (including previously studied polymorphisms in SLC6A4 gene) was analyzed using a dominant model for the risk alleles, the genotypes L/S_10/12_G/A (OR=3.57(95%CI=1.43-8.93); p=0.004, adjusted p-value=0.01) in SLC6A4 and BDNF genes and L/S_10/12_T/C_3/3_G/A in SLC6A4, HTR2A, APOE and BDNF genes (OR=5.99(95%CI=1.66-21.56); p=0.002, adjusted p-value=0.07), were more prevalent in patients than in controls (20%vs.6% and 15%vs.3%, respectively). Even though it is necessary to replicate these findings in a larger population, our results suggest a possible interaction between molecules involved in neurogenesis (BDNF and APOE), serotoninergic neurotransmission (SLC6A4 and HTR2A) and the pathogenesis of MDD. (AU)


La disfunción en la neurotransmisión serotoninérgica ha sido clásicamente asociada con el trastorno depresivo mayor (TDM); sin embargo, otras vías y procesos parecerían tener un rol en esta enfermedad, como la neurogénesis y moléculas asociadas: el factor neurotrófico derivado del cerebro (BDNF) y la apoliproteína E (APOE). Existen reportes en los que se establecen asociaciones entre polimorfismos en estos genes y el TDM. El objetivo de nuestro trabajo fue analizar la posible asociación entre el TDM y polimorfismos en los genes HTR2A (receptor 5-hidroxitriptamina 2A), BDNF y APOE en una muestra de la población argentina previamente estudiada para 2 polimorfismos en el gen SLC6A4 (transportador soluble familia 6 miembro 4). Se estudiaron 5 polimorfismos (rs6311 y rs6313 en HTR2A; rs429358 y rs7412 en APOE; rs6265 en BDNF) en 95 pacientes con TDM y 107 controles no relacionados. No se observaron diferencias significativas entre grupos al analizar la asociación por regresión logística con un único marcador; cuando se analizó el posible efecto combinatorio de polimorfismos (incluyendo los previamente estudiados para el gen SCL6A4) usando un modelo dominante para los alelos de riesgo, los genotipos L/S_10/12_G/A (OR=3,57(95%CI=1,43-8,93); p=0,004, valor-p-ajustado=0,01) en SLC6A4 y BDNF y L/S_10/12_T/C_3/3_G/A en SLC6A4, HTR2A, APOE y BDNF (OR=5,99(95%CI=1,66-21,56); p=0,002, valor-p-ajustado=0,07), fueron más prevalentes en pacientes que controles (20%vs.6% y 15%vs.3% respectivamente). Si bien es necesario replicar estos hallazgos en una población más grande, nuestros resultados sugieren una posible interacción entre moléculas involucradas en la neurogénesis (BDNF y APOE), la neurotransmisión serotoninérgica (SLC6A4 y HTR2A) y la patogenia de la depresión mayor. (AU)


Assuntos
Humanos , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Adulto Jovem , Apolipoproteínas E/deficiência , Polimorfismo Genético , Fator Neurotrófico Derivado do Encéfalo/deficiência , Receptores 5-HT2 de Serotonina/deficiência , Transtorno Depressivo Maior/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/deficiência , Apolipoproteínas E/genética , Argentina/epidemiologia , Fator Neurotrófico Derivado do Encéfalo/genética , Receptores 5-HT2 de Serotonina/genética , Transtorno Depressivo Maior/patologia , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética
15.
Kaohsiung J Med Sci ; 34(3): 134-141, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29475460

RESUMO

The intestinal mucosal barrier (IMB) enables the intestine to provide adequate containment of luminal microorganisms and molecules while preserving the ability to absorb nutrients. In this study, we explored the effect of brain-derived neurotrophic factor (BDNF) on IMB function and gut microbiota in mice. BDNF gene knock-out mice (the BDNF+/- group) and wild-type mice (the BDNF+/+ group) were selected. The gut microbiota of these mice was analyzed by denaturing gradient gel electrophoresis (DGGE) assay. The ultrastructure of the ileum and the colonic epithelium obtained from decapitated mice were observed by transmission electron microscopy. The protein expression of epithelial tight junction proteins, zonula occludens-1 (ZO-1) and occludin was detected by immunohistochemistry staining. The protein expression of claudin-1 and claudin-2 was determined by Western blotting. The DGGE band patterns of gut microbiota in the BDNF+/- group were significantly different from that in the BDNF+/+ group, which indicated that the BDNF expression alters the gut microbiota in mice. Compared with the BDNF+/+ group, the BDNF+/- group presented no significant difference in the ultrastructure of ileal epithelium; however, a significant difference was observed in the colonic epithelial barrier, manifested by decreased microvilli, widening intercellular space and bacterial invasion. Compared with the BDNF+/+ group, the expression of ZO-1 and occludin in the BDNF+/- group was significantly decreased. The expression of claudin-1 in the BDNF+/- group was significantly reduced, while the expression of claudin-2 was elevated. These findings indicate that BDNF preserves IMB function and modulates gut microbiota in mice.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Colo/microbiologia , Microbioma Gastrointestinal/fisiologia , Íleo/microbiologia , Mucosa Intestinal/microbiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/deficiência , Claudina-1/genética , Claudina-1/metabolismo , Claudina-2/genética , Claudina-2/metabolismo , Colo/metabolismo , Colo/ultraestrutura , Regulação da Expressão Gênica , Íleo/metabolismo , Íleo/ultraestrutura , Mucosa Intestinal/metabolismo , Mucosa Intestinal/ultraestrutura , Masculino , Camundongos , Camundongos Knockout , Microvilosidades/metabolismo , Microvilosidades/microbiologia , Microvilosidades/ultraestrutura , Ocludina/genética , Ocludina/metabolismo , Junções Íntimas/metabolismo , Junções Íntimas/microbiologia , Junções Íntimas/ultraestrutura , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
16.
Mol Neurobiol ; 55(3): 2696-2711, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28421542

RESUMO

Traumatic brain injury (TBI) induces cognitive impairments, motor and behavioral deficits. Previous evidences have suggested that neural stem cell (NSC) transplantation could facilitate functional recovery from brain insults, but their underlying mechanisms remains to be elucidated. Here, we established TBI model by an electromagnetic-controlled cortical impact device in the rats. Then, 5 µl NSCs (5.0 × 105/µl), derived from green fluorescent protein (GFP) transgenic mouse, was transplanted into the traumatic brain regions of rats at 24 h after injury. After differentiation of the NSCs was determined using immunohistochemistry, neurological severity scores (NSS) and rotarod test were conducted to detect the neurological behavior. Western blot and RT-PCR as well as ELASA were used to evaluate the expression of synaptophysin and brain-derived neurotrophic factor (BDNF). In order to elucidate the role of BDNF on the neural recovery after NSC transplantation, BDNF knockdown in NSC was performed and transplanted into the rats with TBI, and potential mechanism for BDNF knockdown in the NSC was analyzed using microassay analysis. Meanwhile, BDNF antibody blockade was conducted to further confirm the effect of BDNF on neural activity. As a result, an increasing neurological function improvement was seen in NSC transplanted rats, which was associated with the upregulation of synaptophysin and BDNF expression. Moreover, transplantation of BDNF knockdown NSCs and BDNF antibody block reduced not only the level of synaptophysin but also exacerbated neurological function deficits. Microassay analysis showed that 14 genes such as Wnt and Gsk3-ß were downregulated after BDNF knockdown. The present data therefore showed that BDNF-mediated neuroplasticity underlie the mechanism of NSC transplantation for the treatment of TBI in adult rats.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/terapia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Plasticidade Neuronal/fisiologia , Transplante de Células-Tronco/métodos , Animais , Fator Neurotrófico Derivado do Encéfalo/deficiência , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/fisiologia
17.
Cereb Cortex ; 28(4): 1195-1208, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28184413

RESUMO

Brain-derived neurotrophic factor (BDNF) heterozygous knockout mice (BDNF+/- mice) show fear learning deficits from 3 months of age onwards. Here, we addressed the question how this learning deficit correlates with altered long-term potentiation (LTP) in the cortical synaptic input to the lateral amygdala (LA) and at downstream intra-amygdala synapses in BDNF+/- mice. Our results reveal that the fear learning deficit in BDNF+/- mice was not paralleled by a loss of LTP, neither at cortical inputs to the LA nor at downstream intra-amygdala glutamatergic synapses. As we did observe early fear memory (30 min after training) in BDNF+/- mice while long-term memory (24 h post-training) was absent, the stable LTP in cortico-LA and downstream synapses is in line with the intact acquisition of fear memories. Ex vivo recordings in acute slices of fear-conditioned wildtype (WT) mice revealed that fear learning induces long-lasting changes at cortico-LA synapses that occluded generation of LTP 4 and 24 h after training. Overall, our data show that the intact LTP in the tested amygdala circuits is consistent with intact acquisition of fear memories in both WT and BDNF+/- mice. In addition, the lack of learning-induced long-term changes at cortico-LA synapses in BDNF+/- mice parallels the observed deficit in fear memory consolidation.


Assuntos
Tonsila do Cerebelo/citologia , Fator Neurotrófico Derivado do Encéfalo/deficiência , Medo/fisiologia , Ácido Glutâmico/metabolismo , Aprendizagem/fisiologia , Potenciação de Longa Duração/genética , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Carbazóis/farmacologia , Córtex Cerebral/fisiologia , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Potenciais Evocados/genética , Antagonistas GABAérgicos/farmacologia , Alcaloides Indólicos/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/citologia , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Piridazinas/farmacologia
18.
Mol Cell Neurosci ; 86: 41-49, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29122705

RESUMO

Recent results indicate that STriatal-Enriched protein tyrosine Phosphatase (STEP) levels are regulated by brain-derived neurotrophic factor (BDNF), whose expression changes during postnatal development and aging. Here, we studied STEP ontogeny in mouse brain and changes in STEP with age with emphasis on the possible regulation by BDNF. We found that STEP expression increased during the first weeks of life, reaching adult levels by 2-3weeks of age in the striatum and cortex, and by postnatal day (P) 7 in the hippocampus. STEP protein levels were unaffected in BDNF+/- mice, but were significantly reduced in the striatum and cortex, but not in the hippocampus, of BDNF-/- mice at P7 and P14. In adult wild-type mice there were no changes in cortical and hippocampal STEP61 levels with age. Conversely, striatal STEP levels were reduced from 12months of age, correlating with higher ubiquitination and increased BDNF content and signaling. Lower STEP levels in older mice were paralleled by increased phosphorylation of its substrates. Since altered STEP levels are involved in cellular malfunctioning events, its reduction in the striatum with increasing age should encourage future studies of how this imbalance might participate in the aging process.


Assuntos
Envelhecimento/metabolismo , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Corpo Estriado/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/deficiência , Corpo Estriado/crescimento & desenvolvimento , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
19.
J Neurochem ; 143(1): 49-64, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28722769

RESUMO

Brain-derived neurotrophic factor (BDNF) promotes maturation of cholinergic neurons. However, how activity-dependent BDNF expression affects specific cholinergic gene expression remains unclear. This study addressed this question by determining mRNA levels of 22 acetylcholine receptor subunits, the choline transporter (CHT), and the choline acetyltransferase (ChAT) in mice deficient in activity-dependent BDNF via promoter IV (KIV) and control wild-type mice. Quantitative RT-PCR revealed significant reductions in nicotinic acetylcholine receptor alpha 5 (CHRNA5) in the frontal cortex and hippocampus and M5 muscarinic acetylcholine receptor (CHRM5) in the hippocampus, but significant increases in M2 muscarinic acetylcholine receptor (CHRM2) in the frontal cortex of KIV mice compared to wild-type mice. Three-week treatments with fluoxetine, phenelzine, duloxetine, imipramine, or an enriched environment treatment (EET) did not affect the altered expression of these genes except that EET increased CHRNA5 levels only in KIV frontal cortex. EET also increased levels of CHRNA7, CHT, and ChAT, again only in the KIV frontal cortex. The imipramine treatment was most prominent among the four antidepressants; it up-regulated hippocampal CHRM2 and frontal cortex CHRM5 in both genotypes, and frontal cortex CHRNA7 only in KIV mice. To the best of our knowledge, this is the first evidence that BDNF deficiency disturbs expression of CHRNA5, CHRM2, and CHRM5. Our results suggest that promoter IV-BDNF deficiency - which occurs under chronic stress - causes cholinergic dysfunctions via these receptors. EET is effective on CHRNA5, while its compensatory induction of other cholinergic genes or drugs targeting CHRNA5, CHRM2, and CHRM5 may become an alternative strategy to reverse these BDNF-linked cholinergic dysfunctions.


Assuntos
Antidepressivos/farmacologia , Fator Neurotrófico Derivado do Encéfalo/deficiência , Meio Ambiente , Receptor Muscarínico M2/biossíntese , Receptor Muscarínico M5/biossíntese , Receptores Nicotínicos/biossíntese , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Feminino , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Expressão Gênica , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/fisiologia , Receptor Muscarínico M2/genética , Receptor Muscarínico M5/genética , Receptores Nicotínicos/genética
20.
Behav Brain Res ; 332: 154-163, 2017 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-28576309

RESUMO

Brain derived neurotrophic factor (BDNF) plays a pivotal role in structural plasticity, learning, and memory. Electroencephalogram (EEG) spectral power in the cortex and hippocampus has also been correlated with learning and memory. In this study, we investigated the effect of globally reduced BDNF levels on learning behavior and EEG power via BDNF heterozygous (KO) rats. We employed several behavioral tests that are thought to depend on cortical and hippocampal plasticity to varying degrees: novel object recognition, a test that is reliant on a variety of cognitive systems; contextual fear, which is highly hippocampal-dependent; and cued fear, which has been shown to be amygdala-dependent. We also examined the effects of BDNF reduction on cortical and hippocampal EEG spectral power via chronically implanted electrodes in the motor cortex and dorsal hippocampus. We found that BDNF KO rats were impaired in novelty recognition and fear memory retention, while hippocampal EEG power was decreased in slow waves and increased in fast waves. Interestingly, our results, for the first time, show sexual dimorphism in each of our tests. These results support the hypothesis that BDNF drives both cognitive plasticity and coordinates EEG activity patterns, potentially serving as a link between the two.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/deficiência , Hipocampo/fisiopatologia , Aprendizagem/fisiologia , Transtornos da Memória/fisiopatologia , Córtex Motor/fisiopatologia , Caracteres Sexuais , Animais , Ondas Encefálicas/fisiologia , Fator Neurotrófico Derivado do Encéfalo/genética , Eletrocorticografia , Comportamento Exploratório/fisiologia , Medo/fisiologia , Feminino , Heterozigoto , Masculino , Ratos Sprague-Dawley , Ratos Transgênicos , Reconhecimento Psicológico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...